Pub Date : 2024-10-28eCollection Date: 2024-01-01DOI: 10.1177/17588359241292264
Heqi Yang, Linjuan Li, Xiaofen Li, Yuhang Ma, Yu Yang, Dan Cao
Background: Cholangiocarcinoma is a kind of malignant tumor that originates in the epithelium of the biliary tract. Although there are several options for second-line treatment for patients without specific genetic mutations, the overall treatment efficacy is disappointing. Second-line treatment which is composed of liposomal irinotecan plus fluorouracil and leucovorin significantly improved the treatment efficacy for advanced biliary tract cancer and extended patient survival. This study aims to evaluate the efficacy and safety of the combination of cadonilimab with liposomal irinotecan plus fluorouracil and leucovorin for advanced biliary tract cancer.
Objectives: The primary objective of this study is to determine the objective response rate. The second objectives of this study are overall survival, progression-free survival, disease control rate, and adverse event incidence rate.
Design: The study is a single-arm, prospective phase II clinical trial. In all, 51 patients who are diagnosed with locally advanced or metastatic bile tract cancer will be enrolled.
Methods and analysis: Eligible participants will receive cadonilimab at a dosage of 6 mg/kg on day 1 of each 21-day cycle combined with intravenous liposomal irinotecan at a dosage of 70 mg/m2 for 90 min on day 1 plus leucovorin at a dosage of 400 mg/m2 for 30 min on day 1 and fluorouracil at a dosage of 400 mg/m2 for 46 h every 2 weeks.
Discussion: Previous studies have suggested that there is a synergistic effect between the two treatment modalities. However, the potential of cadonilimab in bile tract cancer has not been explored. Hence, this trial is the first to investigate its efficacy and toxicity. In addition, the trial is also willing to explore potential biomarkers in patients with locally advanced and metastatic bile tract cancer.
Trial registration: This study was registered on ClinicalTrials.gov with NCT06438822.
Ethics: This study protocol and amendments have been approved by the Ethics Committee of West China Hospital (2024(791)).
{"title":"Second-line treatment of PD-1 and CTLA-4 blockade combined with liposomal irinotecan plus leucovorin and fluorouracil for advanced cholangiocarcinoma: study protocol of a single-arm, prospective phase II trial.","authors":"Heqi Yang, Linjuan Li, Xiaofen Li, Yuhang Ma, Yu Yang, Dan Cao","doi":"10.1177/17588359241292264","DOIUrl":"10.1177/17588359241292264","url":null,"abstract":"<p><strong>Background: </strong>Cholangiocarcinoma is a kind of malignant tumor that originates in the epithelium of the biliary tract. Although there are several options for second-line treatment for patients without specific genetic mutations, the overall treatment efficacy is disappointing. Second-line treatment which is composed of liposomal irinotecan plus fluorouracil and leucovorin significantly improved the treatment efficacy for advanced biliary tract cancer and extended patient survival. This study aims to evaluate the efficacy and safety of the combination of cadonilimab with liposomal irinotecan plus fluorouracil and leucovorin for advanced biliary tract cancer.</p><p><strong>Objectives: </strong>The primary objective of this study is to determine the objective response rate. The second objectives of this study are overall survival, progression-free survival, disease control rate, and adverse event incidence rate.</p><p><strong>Design: </strong>The study is a single-arm, prospective phase II clinical trial. In all, 51 patients who are diagnosed with locally advanced or metastatic bile tract cancer will be enrolled.</p><p><strong>Methods and analysis: </strong>Eligible participants will receive cadonilimab at a dosage of 6 mg/kg on day 1 of each 21-day cycle combined with intravenous liposomal irinotecan at a dosage of 70 mg/m<sup>2</sup> for 90 min on day 1 plus leucovorin at a dosage of 400 mg/m<sup>2</sup> for 30 min on day 1 and fluorouracil at a dosage of 400 mg/m<sup>2</sup> for 46 h every 2 weeks.</p><p><strong>Discussion: </strong>Previous studies have suggested that there is a synergistic effect between the two treatment modalities. However, the potential of cadonilimab in bile tract cancer has not been explored. Hence, this trial is the first to investigate its efficacy and toxicity. In addition, the trial is also willing to explore potential biomarkers in patients with locally advanced and metastatic bile tract cancer.</p><p><strong>Trial registration: </strong>This study was registered on ClinicalTrials.gov with NCT06438822.</p><p><strong>Ethics: </strong>This study protocol and amendments have been approved by the Ethics Committee of West China Hospital (2024(791)).</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241292264"},"PeriodicalIF":4.3,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11528739/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142569710","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Increasingly, more evidence has shown that inflammation stress and the tumor microenvironment pose a negative effect on targeted therapy. The neutrophil-to-lymphocyte ratio is considered to be a surrogate biomarker of inflammation and can predict pazopanib treatment effect in non-adipocytic soft-tissue sarcoma (STS). The role of the pan-immune-inflammation value (PIV) in STS is still yet to be determined.
Objectives: We sought whether the pre-treatment PIV could be applied to predict the response of pazopanib in STS.
Design: We conducted a retrospective analysis of 75 patients who had been treated with pazopanib for recurrent or metastatic non-adipocytic STS.
Methods: Our cohort was stratified into either a pre-treatment high PIV group with PIV ⩾310 (n = 45) or a low PIV group with PIV <310 (n = 30). We compared their clinical features and outcomes. Cox regression analysis was employed to determine the risk factors of disease progression and mortality. Kaplan-Meier survival curves were utilized to assess both the progression-free survival (PFS) and overall survival (OS).
Results: The results revealed that a pre-treatment high PIV (⩾310) is a risk factor for progression under pazopanib (hazard ratio: 1.91; 95% confidence interval: 1.08-3.36; p = 0.025). The median PFS and OS of the pre-treatment high PIV group were found to be significantly lower than the low PIV group (0.33 vs 0.75 years; p = 0.023, 0.46 vs 1.63 years; p = 0.025).
Conclusion: High pre-treatment PIV in STS patients may indicate an elevated risk of disease progression and mortality. Pre-treatment PIV reflects inflammation stress and acts as a practical biomarker for STS patients treated with pazopanib.
背景:越来越多的证据表明,炎症应激和肿瘤微环境会对靶向治疗产生负面影响。中性粒细胞与淋巴细胞比值被认为是炎症的替代生物标志物,可以预测非脂肪细胞软组织肉瘤(STS)中帕唑帕尼的治疗效果。泛免疫炎症值(PIV)在STS中的作用仍有待确定:我们试图了解治疗前 PIV 是否可用于预测帕唑帕尼对 STS 的反应:我们对75例接受帕唑帕尼治疗的复发性或转移性非脂肪细胞STS患者进行了回顾性分析:我们将患者分为治疗前高PIV组(PIV ⩾310)(n = 45)或低PIV组(PIV n = 30)。我们比较了他们的临床特征和预后。我们采用了 Cox 回归分析来确定疾病进展和死亡率的风险因素。卡普兰-梅耶生存曲线用于评估无进展生存期(PFS)和总生存期(OS):结果显示,治疗前高 PIV(⩾310)是帕唑帕尼治疗进展的危险因素(危险比:1.91;95% 置信区间:1.08-3.36;P = 0.025)。治疗前高PIV组的中位PFS和OS明显低于低PIV组(0.33 vs 0.75 years; p = 0.023,0.46 vs 1.63 years; p = 0.025):结论:STS患者治疗前的高PIV可能预示着疾病进展和死亡风险的升高。治疗前PIV反映了炎症应激,是接受帕唑帕尼治疗的STS患者的实用生物标志物。
{"title":"Pre-treatment pan-immune-inflammation value as a prognostic marker of pazopanib in soft tissue sarcoma.","authors":"Cheng-Han Wu, Cheng-Lun Lai, Yong-Chen Hsu, Chiann-Yi Hsu, Yu-Chao Wang, Hsin-Chen Lin","doi":"10.1177/17588359241292255","DOIUrl":"10.1177/17588359241292255","url":null,"abstract":"<p><strong>Background: </strong>Increasingly, more evidence has shown that inflammation stress and the tumor microenvironment pose a negative effect on targeted therapy. The neutrophil-to-lymphocyte ratio is considered to be a surrogate biomarker of inflammation and can predict pazopanib treatment effect in non-adipocytic soft-tissue sarcoma (STS). The role of the pan-immune-inflammation value (PIV) in STS is still yet to be determined.</p><p><strong>Objectives: </strong>We sought whether the pre-treatment PIV could be applied to predict the response of pazopanib in STS.</p><p><strong>Design: </strong>We conducted a retrospective analysis of 75 patients who had been treated with pazopanib for recurrent or metastatic non-adipocytic STS.</p><p><strong>Methods: </strong>Our cohort was stratified into either a pre-treatment high PIV group with PIV ⩾310 (<i>n</i> = 45) or a low PIV group with PIV <310 (<i>n</i> = 30). We compared their clinical features and outcomes. Cox regression analysis was employed to determine the risk factors of disease progression and mortality. Kaplan-Meier survival curves were utilized to assess both the progression-free survival (PFS) and overall survival (OS).</p><p><strong>Results: </strong>The results revealed that a pre-treatment high PIV (⩾310) is a risk factor for progression under pazopanib (hazard ratio: 1.91; 95% confidence interval: 1.08-3.36; <i>p</i> = 0.025). The median PFS and OS of the pre-treatment high PIV group were found to be significantly lower than the low PIV group (0.33 vs 0.75 years; <i>p</i> = 0.023, 0.46 vs 1.63 years; <i>p</i> = 0.025).</p><p><strong>Conclusion: </strong>High pre-treatment PIV in STS patients may indicate an elevated risk of disease progression and mortality. Pre-treatment PIV reflects inflammation stress and acts as a practical biomarker for STS patients treated with pazopanib.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241292255"},"PeriodicalIF":4.3,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11523153/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142547631","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-23eCollection Date: 2024-01-01DOI: 10.1177/17588359241290482
Min Kyu Kim, In Rae Cho, Yooeun Kim, Jin Ho Choi, Kwangrok Jung, Jaihwan Kim, Sheehyun Kim, Hongseok Yun, Jeesun Yoon, Do-Youn Oh, Kwangsoo Kim, Sang Hyub Lee
Background: KRAS, TP53, CDKN2A, and SMAD4 have been the main driver mutations in pancreatic ductal adenocarcinoma (PDAC). Studies on the clinical significance and treatment response to 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) regimen in terms of the presence of these mutations remain inconclusive.
Objectives: This study aimed to compare the survival outcome and response to FOLFIRINOX chemotherapy based on the presence of four driver mutation genes.
Design: A multi-center retrospective study conducted at two tertiary medical centers.
Methods: This study analyzed PDAC patients who were treated with FOLFIRINOX chemotherapy as the initial treatment. Tumor specimens were analyzed by a targeted next-generation sequencing platform at two tertiary referral hospitals from January 2016 to March 2022. Patients' demographics, survival outcomes, and chemotherapeutic response were investigated and compared according to the presence of driver mutations.
Results: The analysis included 100 patients. KRAS mutation was identified in 92 (92.0%) patients, followed by TP53, CDKN2A, and SMAD4 in 63 (63.0%), 18 (18.0%), and 17 (17.0%) patients, respectively. The TP53 wild-type group demonstrated longer overall survival (OS) than the TP53 mutated group (median OS: 29 vs 19 months, p = 0.03), and TP53 served as a prognostic factor for survival (hazard ratio = 1.74, 95% confidence interval: 1.00-3.00, p = 0.048). The difference in OS according to TP53 mutation was intensified in localized pancreatic adenocarcinoma (37 vs 19 months, p = 0.01). The TP53 wild-type group demonstrated a higher objective response rate to FOLFIRINOX chemotherapy than the TP53 mutation group in localized pancreatic adenocarcinoma (50.0% vs 17.6%, p = 0.024).
Conclusion: PDAC patients with wild-type TP53 demonstrated longer OS than those with TP53 mutation, and this trend was intensified in patients with localized disease. This result may be due to an impaired response to FOLFIRINOX chemotherapy in patients with TP53 mutation.
背景:KRAS、TP53、CDKN2A和SMAD4是胰腺导管腺癌(PDAC)的主要驱动突变。关于这些突变存在的临床意义和对 5-氟尿嘧啶、亮菌素、伊立替康和奥沙利铂(FOLFIRINOX)方案的治疗反应的研究仍无定论:本研究旨在根据四种驱动突变基因的存在情况,比较 FOLFIRINOX 化疗的生存结果和反应:在两家三级医疗中心开展的多中心回顾性研究:本研究分析了接受FOLFIRINOX化疗作为初始治疗的PDAC患者。2016年1月至2022年3月期间,两家三级转诊医院通过靶向新一代测序平台对肿瘤标本进行了分析。根据是否存在驱动基因突变,对患者的人口统计学特征、生存结果和化疗反应进行了调查和比较:分析包括 100 名患者。92例(92.0%)患者发现了KRAS突变,63例(63.0%)、18例(18.0%)和17例(17.0%)患者发现了TP53、CDKN2A和SMAD4突变。TP53 野生型组的总生存期(OS)长于 TP53 突变组(中位 OS:29 个月 vs 19 个月,P = 0.03),TP53 是生存期的预后因素(危险比 = 1.74,95% 置信区间:1.00-3.00,P = 0.048)。在局部胰腺癌中,TP53突变导致的生存期差异更大(37个月 vs 19个月,p = 0.01)。在局部胰腺腺癌中,TP53野生型组对FOLFIRINOX化疗的客观反应率高于TP53突变组(50.0% vs 17.6%,P = 0.024):结论:TP53野生型的PDAC患者的OS比TP53突变的患者长,这一趋势在局部疾病患者中更加明显。这一结果可能是由于TP53突变患者对FOLFIRINOX化疗的反应减弱所致。
{"title":"Prognostic value of the <i>TP53</i> mutation in patients with pancreatic ductal adenocarcinoma receiving FOLFIRINOX.","authors":"Min Kyu Kim, In Rae Cho, Yooeun Kim, Jin Ho Choi, Kwangrok Jung, Jaihwan Kim, Sheehyun Kim, Hongseok Yun, Jeesun Yoon, Do-Youn Oh, Kwangsoo Kim, Sang Hyub Lee","doi":"10.1177/17588359241290482","DOIUrl":"https://doi.org/10.1177/17588359241290482","url":null,"abstract":"<p><strong>Background: </strong><i>KRAS</i>, <i>TP53</i>, <i>CDKN2A</i>, and <i>SMAD4</i> have been the main driver mutations in pancreatic ductal adenocarcinoma (PDAC). Studies on the clinical significance and treatment response to 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) regimen in terms of the presence of these mutations remain inconclusive.</p><p><strong>Objectives: </strong>This study aimed to compare the survival outcome and response to FOLFIRINOX chemotherapy based on the presence of four driver mutation genes.</p><p><strong>Design: </strong>A multi-center retrospective study conducted at two tertiary medical centers.</p><p><strong>Methods: </strong>This study analyzed PDAC patients who were treated with FOLFIRINOX chemotherapy as the initial treatment. Tumor specimens were analyzed by a targeted next-generation sequencing platform at two tertiary referral hospitals from January 2016 to March 2022. Patients' demographics, survival outcomes, and chemotherapeutic response were investigated and compared according to the presence of driver mutations.</p><p><strong>Results: </strong>The analysis included 100 patients. <i>KRAS</i> mutation was identified in 92 (92.0%) patients, followed by <i>TP53</i>, <i>CDKN2A</i>, and <i>SMAD4</i> in 63 (63.0%), 18 (18.0%), and 17 (17.0%) patients, respectively. The <i>TP53</i> wild-type group demonstrated longer overall survival (OS) than the <i>TP53</i> mutated group (median OS: 29 vs 19 months, <i>p</i> = 0.03), and <i>TP53</i> served as a prognostic factor for survival (hazard ratio = 1.74, 95% confidence interval: 1.00-3.00, <i>p</i> = 0.048). The difference in OS according to <i>TP53</i> mutation was intensified in localized pancreatic adenocarcinoma (37 vs 19 months, <i>p</i> = 0.01). The <i>TP53</i> wild-type group demonstrated a higher objective response rate to FOLFIRINOX chemotherapy than the <i>TP53</i> mutation group in localized pancreatic adenocarcinoma (50.0% vs 17.6%, <i>p</i> = 0.024).</p><p><strong>Conclusion: </strong>PDAC patients with wild-type <i>TP53</i> demonstrated longer OS than those with <i>TP53</i> mutation, and this trend was intensified in patients with localized disease. This result may be due to an impaired response to FOLFIRINOX chemotherapy in patients with <i>TP53</i> mutation.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241290482"},"PeriodicalIF":4.3,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11500227/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142508536","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-23eCollection Date: 2024-01-01DOI: 10.1177/17588359241290720
Anna Pous, Adrià Bernat-Peguera, Assumpció López-Paradís, Beatriz Cirauqui, Vanesa Quiroga, Iris Teruel, Eudald Felip, Angelica Ferrando-Díez, Milana Bergamino, Laia Boronat, Margarita Romeo, Gemma Soler, Christian Mariño, Paula Rodríguez-Martínez, Laura Pons, Ester Ballana, Anna Martinez-Cardús, Mireia Margelí
Background: Human epidermal growth factor receptor 2 (HER2)-low has emerged as a potential new entity in breast cancer (BC). Data on this subset are limited, and prognostic results are controversial, evidencing the need of further data in a BC real-world cohort.
Methods: Patients with HER2-negative stage I-III BC diagnosed between 2006 and 2016 were retrospectively reviewed in a single cohort from the Catalan Institute of Oncology Badalona. Demographics and clinicopathological characteristics were examined via medical charts/electronic health records. We aim to describe and compare HER2-0/HER2-low populations through Chi-square or Fisher test, and explore its prognostic impact using Kaplan-Meier curves and Cox regression models.
Results: From a cohort of 1755 BC patients, 1401 invasive HER2-negative, stage I-III cases were evaluated. 87% were hormone receptor (HR)-positive versus 13% triple negative (TNBC). Overall, 43% were HER2-0 and 57% HER2-low (61% immunohistochemistry (IHC) 1+ and 39% IHC 2+). Comparing HER2-low versus HER2-0, HER2-low showed higher proportion of estrogen receptor (ER)-positive (91.6% vs 79.9%, p ⩽ 0.001) and progesterone receptor (PR)-positive (79.8% vs 68.9%, p ⩽ 0.001) cases. HER2-0 exhibited higher proportion of TNBC (20.1% vs 8.4%, p = 0.001), grade III tumors (28.8% vs 23.5%, p = 0.039), and higher Ki67 median value (26.47% vs 23.88%, p = 0.041). HER2-low was associated with longer time to distant recurrence (TTDR) compared to HER2-0 (67.8 vs 54.1 months; p = 0.015) and better BC-related survival (19.2 vs 16.3 years; p = 0.033). In the multivariable analysis, HER2-low was not an independent prognostic factor for TTDR and BC-related survival. ER expression showed a strong association with longer TTDR (Hazard Ratio: 0.425, p ⩽ 0.001) and improved BC-related survival (Hazard Ratio: 0.380, p ⩽ 0.001). PR expression was also associated with longer TTDR (Hazard Ratio: 0.496, p ⩽ 0.001), and improved BC-related survival (Hazard Ratio: 0.488, p ⩽ 0.001). Histological grade III was significantly associated with shorter TTDR (Hazard Ratio: 1.737, p = 0.002). Positive nodal status was the strongest factor correlated with worse BC-related survival (Hazard Ratio: 2.747, p ⩽ 0.001).
Conclusion: HER2-low was significantly associated with HR-positive disease, whereas HER2-0 group had higher incidence of TNBC, histological grade III and higher Ki67%. Although HER2-low group was associated with longer TTDR and improved BC-related survival, these findings could be explained by the greater proportion of favorable prognostic features in this subgroup compared to HER2-0.
背景:低人类表皮生长因子受体2(HER2)已成为乳腺癌(BC)的一个潜在新实体。有关这一亚群的数据有限,预后结果也存在争议,这表明需要在乳腺癌真实世界队列中获得更多数据:方法:对加泰罗尼亚巴达洛纳肿瘤研究所(Catalan Institute of Oncology Badalona)的单一队列中2006年至2016年间确诊的HER2阴性I-III期BC患者进行回顾性研究。通过病历/电子病历研究了人口统计学和临床病理学特征。我们旨在通过Chi-square或Fisher检验对HER2-0/HER2-low人群进行描述和比较,并通过Kaplan-Meier曲线和Cox回归模型探讨其对预后的影响:在 1755 例 BC 患者中,评估了 1401 例侵袭性 HER2 阴性、I-III 期病例。87%为激素受体(HR)阳性,13%为三阴性(TNBC)。总体而言,43%为HER2-0,57%为HER2-低(61%为免疫组化(IHC)1+,39%为免疫组化(IHC)2+)。HER2 低与 HER2-0 相比,HER2-低的病例中雌激素受体 (ER) 阳性(91.6% 对 79.9%,P ⩽0.001)和孕激素受体 (PR) 阳性(79.8% 对 68.9%,P ⩽0.001)的比例更高。HER2-0表现出更高的TNBC比例(20.1% vs 8.4%,p = 0.001)、III级肿瘤(28.8% vs 23.5%,p = 0.039)和更高的Ki67中位值(26.47% vs 23.88%,p = 0.041)。与HER2-0相比,HER2-低与更长的远处复发时间(TTDR)(67.8个月 vs 54.1个月;p = 0.015)和更好的BC相关生存期(19.2年 vs 16.3年;p = 0.033)相关。在多变量分析中,HER2-低并不是TTDR和BC相关生存期的独立预后因素。ER表达与更长的TTDR(危险比:0.425,p ⩽0.001)和更高的BC相关生存率(危险比:0.380,p ⩽0.001)密切相关。PR 表达也与较长的 TTDR 相关(危险比:0.496,p ⩽0.001),并能改善 BC 相关生存率(危险比:0.488,p ⩽0.001)。组织学 III 级与较短的 TTDR 显著相关(危险比:1.737,p = 0.002)。结节状态阳性是与较差的 BC 相关生存率相关的最强因素(危险比:2.747,p ⩽0.001):结论:HER2-低与HR阳性疾病明显相关,而HER2-0组的TNBC发病率更高,组织学分级为III级,Ki67%更高。虽然HER2-低组与更长的TTDR和更高的BC相关生存率有关,但与HER2-0组相比,该亚组中有利预后特征的比例更高,这可能是这些发现的原因。
{"title":"Deciphering HER2-low breast cancer (BC): insights from real-world data in early stage breast cancer.","authors":"Anna Pous, Adrià Bernat-Peguera, Assumpció López-Paradís, Beatriz Cirauqui, Vanesa Quiroga, Iris Teruel, Eudald Felip, Angelica Ferrando-Díez, Milana Bergamino, Laia Boronat, Margarita Romeo, Gemma Soler, Christian Mariño, Paula Rodríguez-Martínez, Laura Pons, Ester Ballana, Anna Martinez-Cardús, Mireia Margelí","doi":"10.1177/17588359241290720","DOIUrl":"https://doi.org/10.1177/17588359241290720","url":null,"abstract":"<p><strong>Background: </strong>Human epidermal growth factor receptor 2 (HER2)-low has emerged as a potential new entity in breast cancer (BC). Data on this subset are limited, and prognostic results are controversial, evidencing the need of further data in a BC real-world cohort.</p><p><strong>Methods: </strong>Patients with HER2-negative stage I-III BC diagnosed between 2006 and 2016 were retrospectively reviewed in a single cohort from the Catalan Institute of Oncology Badalona. Demographics and clinicopathological characteristics were examined via medical charts/electronic health records. We aim to describe and compare HER2-0/HER2-low populations through Chi-square or Fisher test, and explore its prognostic impact using Kaplan-Meier curves and Cox regression models.</p><p><strong>Results: </strong>From a cohort of 1755 BC patients, 1401 invasive HER2-negative, stage I-III cases were evaluated. 87% were hormone receptor (HR)-positive versus 13% triple negative (TNBC). Overall, 43% were HER2-0 and 57% HER2-low (61% immunohistochemistry (IHC) 1+ and 39% IHC 2+). Comparing HER2-low versus HER2-0, HER2-low showed higher proportion of estrogen receptor (ER)-positive (91.6% vs 79.9%, <i>p</i> ⩽ 0.001) and progesterone receptor (PR)-positive (79.8% vs 68.9%, <i>p</i> ⩽ 0.001) cases. HER2-0 exhibited higher proportion of TNBC (20.1% vs 8.4%, <i>p</i> = 0.001), grade III tumors (28.8% vs 23.5%, <i>p</i> = 0.039), and higher Ki67 median value (26.47% vs 23.88%, <i>p</i> = 0.041). HER2-low was associated with longer time to distant recurrence (TTDR) compared to HER2-0 (67.8 vs 54.1 months; <i>p</i> = 0.015) and better BC-related survival (19.2 vs 16.3 years; <i>p</i> = 0.033). In the multivariable analysis, HER2-low was not an independent prognostic factor for TTDR and BC-related survival. ER expression showed a strong association with longer TTDR (Hazard Ratio: 0.425, <i>p</i> ⩽ 0.001) and improved BC-related survival (Hazard Ratio: 0.380, <i>p</i> ⩽ 0.001). PR expression was also associated with longer TTDR (Hazard Ratio: 0.496, <i>p</i> ⩽ 0.001), and improved BC-related survival (Hazard Ratio: 0.488, <i>p</i> ⩽ 0.001). Histological grade III was significantly associated with shorter TTDR (Hazard Ratio: 1.737, <i>p</i> = 0.002). Positive nodal status was the strongest factor correlated with worse BC-related survival (Hazard Ratio: 2.747, <i>p</i> ⩽ 0.001).</p><p><strong>Conclusion: </strong>HER2-low was significantly associated with HR-positive disease, whereas HER2-0 group had higher incidence of TNBC, histological grade III and higher Ki67%. Although HER2-low group was associated with longer TTDR and improved BC-related survival, these findings could be explained by the greater proportion of favorable prognostic features in this subgroup compared to HER2-0.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241290720"},"PeriodicalIF":4.3,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11500235/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142508534","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Abemaciclib was the first cyclin-dependent kinase 4/6 (CDK4/6) inhibitor approved globally in the adjuvant setting for high-risk hormone-receptor positive (HR+)/human epidermal growth factor 2 negative (HER2-) early breast cancer (EBC), based on the phase III monarchE trial.
Objective: To report an exploratory Chinese population analysis based on the preplanned overall survival (OS) interim analysis with 5-year efficacy results of monarchE.
Design and methods: Patients with HR+/HER2-, high-risk (⩾4 positive lymph nodes, or 1-3 nodes and either tumor size ⩾5 cm, histologic grade 3, or Ki-67 ⩾20%) EBC were randomized (1:1) to abemaciclib (150 mg twice daily for 2 years) plus endocrine therapy (ET), or ET alone. This analysis included Chinese patients enrolled in mainland China, Hong Kong, and Taiwan. The primary endpoint was invasive disease-free survival (IDFS); key secondary endpoints included distant relapse-free survival (DRFS), safety, and patient-reported outcomes (PROs).
Results: Overall, 501 Chinese patients were included (abemaciclib + ET, n = 259; ET, n = 242). With a median follow-up of 53 months, the addition of abemaciclib to ET resulted in improvements in IDFS (estimated 5-year IDFS rate: 85.9% vs 79.1%; hazard ratio (HR), 0.65 (95% confidence interval (CI) 0.41-1.03)) and DRFS (estimated 5-year DRFS rate: 88.4% vs 82.3%; HR, 0.65 (95% CI, 0.39-1.07)). The most common grade ⩾3 treatment-emergent adverse events in the abemaciclib + ET versus ET groups were neutropenia (24.7% vs 0.8%) and leukopenia (22.4% vs 0.4%). Generally, no clinically meaningful difference in PROs (endocrine symptoms and fatigue) was observed between groups, except for diarrhea.
Conclusion: At this prespecified OS interim analysis, which provides 5-year data, the addition of abemaciclib to ET in Chinese patients with high-risk HR+, HER2- EBC was associated with sustained and clinically meaningful improvements in IDFS and DRFS, with acceptable safety and tolerability profiles and minimal impact on PROs. These results represent the first full report of a CDK4/6 inhibitor in Chinese patients with EBC and support the positive benefit-risk profile of adjuvant abemaciclib + ET in Chinese patients.
Trial registration: ClinicalTrials.gov identifier: NCT03155997 (first posted: May 16, 2017).
背景基于monarchEⅢ期试验,Abemaciclib是全球首个获批用于高危激素受体阳性(HR+)/人表皮生长因子2阴性(HER2-)早期乳腺癌(EBC)辅助治疗的细胞周期蛋白依赖性激酶4/6(CDK4/6)抑制剂:报告基于monarchE 5年疗效的预计划总生存期(OS)中期分析的探索性中国人群分析:将HR+/HER2-、高危(淋巴结阳性⩾4个,或1-3个淋巴结且肿瘤大小⩾5 cm、组织学分级3级或Ki-67 ⩾20%)EBC患者随机(1:1)分为阿贝单抗(150 mg,每日2次,疗程2年)+内分泌治疗(ET)或单用ET。该分析包括在中国大陆、香港和台湾注册的中国患者。主要终点是无侵袭性疾病生存期(IDFS);关键次要终点包括无远处复发生存期(DRFS)、安全性和患者报告结果(PROs):共纳入501名中国患者(abemaciclib + ET,n = 259;ET,n = 242)。中位随访时间为53个月,在ET基础上加用阿巴西利可改善了IDFS(估计5年IDFS率为85.9% vs 79.1%):85.9% vs 79.1%;危险比 (HR),0.65(95% 置信区间 (CI),0.41-1.03))和 DRFS(估计 5 年 DRFS 率:88.4% vs 82.3%):88.4%对82.3%;HR,0.65(95% 置信区间,0.39-1.07))。阿昔单抗+ET组与ET组相比,最常见的⩾3级治疗突发不良事件是中性粒细胞减少(24.7% vs 0.8%)和白细胞减少(22.4% vs 0.4%)。总体而言,除腹泻外,各组间在PROs(内分泌症状和疲劳)方面未观察到有临床意义的差异:此次预设的OS中期分析提供了5年的数据,在中国高危HR+、HER2- EBC患者的ET基础上加用阿巴西利,可持续改善IDFS和DRFS,且具有临床意义,安全性和耐受性均可接受,对PROs的影响极小。这些结果代表了CDK4/6抑制剂在中国EBC患者中的首次全面报道,并支持中国患者辅助治疗阿巴西利+ET的积极获益-风险特征:试验注册:ClinicalTrials.gov identifier:NCT03155997(首次发布时间:2017年5月16日)。
{"title":"Abemaciclib combined with endocrine therapy as adjuvant treatment for hormone-receptor-positive, HER2-, high-risk early breast cancer: 5-year Chinese population analysis of the phase III randomized monarchE study.","authors":"Qingyuan Zhang, Kunwei Shen, Chuan-Gui Song, Quchang Ouyang, Zhenzhen Liu, Qiang Liu, Jifeng Feng, Joanne W Y Chiu, Jinhai Tang, Zefei Jiang, Ling-Ming Tseng, Xiaojia Wang, Liu Yang, Chenxi Qian, Zhimin Shao","doi":"10.1177/17588359241286775","DOIUrl":"10.1177/17588359241286775","url":null,"abstract":"<p><strong>Background: </strong>Abemaciclib was the first cyclin-dependent kinase 4/6 (CDK4/6) inhibitor approved globally in the adjuvant setting for high-risk hormone-receptor positive (HR+)/human epidermal growth factor 2 negative (HER2-) early breast cancer (EBC), based on the phase III monarchE trial.</p><p><strong>Objective: </strong>To report an exploratory Chinese population analysis based on the preplanned overall survival (OS) interim analysis with 5-year efficacy results of monarchE.</p><p><strong>Design and methods: </strong>Patients with HR+/HER2-, high-risk (⩾4 positive lymph nodes, or 1-3 nodes and either tumor size ⩾5 cm, histologic grade 3, or Ki-67 ⩾20%) EBC were randomized (1:1) to abemaciclib (150 mg twice daily for 2 years) plus endocrine therapy (ET), or ET alone. This analysis included Chinese patients enrolled in mainland China, Hong Kong, and Taiwan. The primary endpoint was invasive disease-free survival (IDFS); key secondary endpoints included distant relapse-free survival (DRFS), safety, and patient-reported outcomes (PROs).</p><p><strong>Results: </strong>Overall, 501 Chinese patients were included (abemaciclib + ET, <i>n</i> = 259; ET, <i>n</i> = 242). With a median follow-up of 53 months, the addition of abemaciclib to ET resulted in improvements in IDFS (estimated 5-year IDFS rate: 85.9% vs 79.1%; hazard ratio (HR), 0.65 (95% confidence interval (CI) 0.41-1.03)) and DRFS (estimated 5-year DRFS rate: 88.4% vs 82.3%; HR, 0.65 (95% CI, 0.39-1.07)). The most common grade ⩾3 treatment-emergent adverse events in the abemaciclib + ET versus ET groups were neutropenia (24.7% vs 0.8%) and leukopenia (22.4% vs 0.4%). Generally, no clinically meaningful difference in PROs (endocrine symptoms and fatigue) was observed between groups, except for diarrhea.</p><p><strong>Conclusion: </strong>At this prespecified OS interim analysis, which provides 5-year data, the addition of abemaciclib to ET in Chinese patients with high-risk HR+, HER2- EBC was associated with sustained and clinically meaningful improvements in IDFS and DRFS, with acceptable safety and tolerability profiles and minimal impact on PROs. These results represent the first full report of a CDK4/6 inhibitor in Chinese patients with EBC and support the positive benefit-risk profile of adjuvant abemaciclib + ET in Chinese patients.</p><p><strong>Trial registration: </strong>ClinicalTrials.gov identifier: NCT03155997 (first posted: May 16, 2017).</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241286775"},"PeriodicalIF":4.3,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11503738/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142508532","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-21eCollection Date: 2024-01-01DOI: 10.1177/17588359241287747
Tanya T D Soeratram, Isis Beentjes, Jacqueline M P Egthuijsen, Aart Mookhoek, Marilyne M Lange, Elma Meershoek-Klein Kranenbarg, Henk H Hartgrink, Cornelis J H van de Velde, Bauke Ylstra, Hanneke W M van Laarhoven, Nicole C T van Grieken
Background: The prognostic significance of T-cell densities in gastric cancer (GC) was previously demonstrated in surgical resection specimens. For prognosis or response prediction, it is preferable to identify biomarkers in pre-treatment biopsies; yet, its representativeness of the tumor immune microenvironment is unclear.
Objectives: This study aimed to evaluate the concordance and prognostic value of T-cell densities in paired biopsies and resections.
Methods: Paired diagnostic biopsies and surgical resections were available for 131 patients with resectable GC who were treated with surgery alone in the D1/D2 trial. T-cell markers such as CD3, CD45RO, CD8, FOXP3, and Granzyme B were assessed by immunohistochemistry and digitally quantified. Tumors were categorized into high and low subgroups for each marker. The concordance between biopsies and resections was determined for each marker with Cohen's κ. To determine the prognostic value of T cells in biopsies, Cox regression was performed.
Results: The concordance of T-cell high and low tumors was moderate for CD8 (κ = 0.58) and weak for other markers (κ < 0.3). CD8 and FOXP3 densities in biopsies were significantly associated with cancer-specific survival. Multivariable analysis showed that an Immunoscore incorporating CD8 and FOXP3 served as an independent prognostic marker (low vs high: hazard ratio 3.40, 95% confidence interval: 1.27-9.10; p = 0.015).
Conclusion: Although the concordance in T-cell densities between biopsy and resection specimens is modest, a biopsy-based Immunoscore identified distinct biological subgroups with prognostic potential. To fully evaluate the prognostic performance of this biopsy Immunoscore, additional studies are warranted.
背景:胃癌(GC)手术切除标本中的T细胞密度具有预后意义。对于预后或反应预测,最好在治疗前活检中确定生物标志物;然而,其对肿瘤免疫微环境的代表性尚不清楚:本研究旨在评估配对活检和切除术中 T 细胞密度的一致性和预后价值:方法:在 D1/D2 试验中,131 名接受单纯手术治疗的可切除 GC 患者接受了配对诊断活检和手术切除。CD3、CD45RO、CD8、FOXP3 和 Granzyme B 等 T 细胞标记物通过免疫组化方法进行评估,并进行数字量化。每种标记物的肿瘤被分为高亚组和低亚组。每个标记物的活检和切除之间的一致性用 Cohen's κ 来确定。为确定活检中 T 细胞的预后价值,进行了 Cox 回归:结果:T细胞含量高和含量低的肿瘤在CD8(κ = 0.58)方面的一致性为中等,而在其他标记物(κ p = 0.015)方面的一致性较弱:尽管活检标本和切除标本的T细胞密度一致性不高,但基于活检的Immunoscore发现了具有预后潜力的不同生物亚群。要全面评估这种活检免疫评分的预后性能,还需要进行更多的研究。
{"title":"A biopsy-based Immunoscore in patients with treatment-naïve resectable gastric cancer.","authors":"Tanya T D Soeratram, Isis Beentjes, Jacqueline M P Egthuijsen, Aart Mookhoek, Marilyne M Lange, Elma Meershoek-Klein Kranenbarg, Henk H Hartgrink, Cornelis J H van de Velde, Bauke Ylstra, Hanneke W M van Laarhoven, Nicole C T van Grieken","doi":"10.1177/17588359241287747","DOIUrl":"https://doi.org/10.1177/17588359241287747","url":null,"abstract":"<p><strong>Background: </strong>The prognostic significance of T-cell densities in gastric cancer (GC) was previously demonstrated in surgical resection specimens. For prognosis or response prediction, it is preferable to identify biomarkers in pre-treatment biopsies; yet, its representativeness of the tumor immune microenvironment is unclear.</p><p><strong>Objectives: </strong>This study aimed to evaluate the concordance and prognostic value of T-cell densities in paired biopsies and resections.</p><p><strong>Methods: </strong>Paired diagnostic biopsies and surgical resections were available for 131 patients with resectable GC who were treated with surgery alone in the D1/D2 trial. T-cell markers such as CD3, CD45RO, CD8, FOXP3, and Granzyme B were assessed by immunohistochemistry and digitally quantified. Tumors were categorized into high and low subgroups for each marker. The concordance between biopsies and resections was determined for each marker with Cohen's κ. To determine the prognostic value of T cells in biopsies, Cox regression was performed.</p><p><strong>Results: </strong>The concordance of T-cell high and low tumors was moderate for CD8 (κ = 0.58) and weak for other markers (κ < 0.3). CD8 and FOXP3 densities in biopsies were significantly associated with cancer-specific survival. Multivariable analysis showed that an Immunoscore incorporating CD8 and FOXP3 served as an independent prognostic marker (low vs high: hazard ratio 3.40, 95% confidence interval: 1.27-9.10; <i>p</i> = 0.015).</p><p><strong>Conclusion: </strong>Although the concordance in T-cell densities between biopsy and resection specimens is modest, a biopsy-based Immunoscore identified distinct biological subgroups with prognostic potential. To fully evaluate the prognostic performance of this biopsy Immunoscore, additional studies are warranted.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241287747"},"PeriodicalIF":4.3,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11497501/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142508531","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-21eCollection Date: 2024-01-01DOI: 10.1177/17588359241280689
Sun Min Lim, Stefanie S Schalm, Eun Ji Lee, Sewon Park, Chiara Conti, Yves A Millet, Rich Woessner, Zhuo Zhang, Luz E Tavera-Mendoza, Faith Stevison, Faris Albayya, Thomas A Dineen, John Hsieh, Seung Yeon Oh, Alena Zalutskaya, Julia Rotow, Koichi Goto, Dae-Ho Lee, Mi Ran Yun, Byoung Chul Cho
Introduction: Despite the availability of several epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs), most patients with non-small-cell lung cancer (NSCLC) eventually develop resistance to these agents. Notably, EGFR_C797S mutations confer resistance to the third-generation EGFR-TKI osimertinib and no approved post-osimertinib targeted pharmacology options are currently available. BLU-945 is a novel, reversible, and orally available next-generation EGFR-TKI that selectively targets EGFR-activating (EGFRm) and resistance mutations (including EGFR_C797S) with nanomolar potency while sparing wild-type EGFR in vitro.
Methods: In vitro activity of BLU-945 as a single agent and in combination with osimertinib was tested in engineered EGFR-mutant cell lines as well as patient-derived cells and patient-derived organoids. In vivo activity was evaluated in osimertinib-resistant patient-derived xenograft mouse models. Three patient cases from the global, first-in-human, phase I/II SYMPHONY trial (NCT04862780) demonstrating the clinical efficacy of BLU-945 were reported.
Results: In vitro BLU-945 demonstrated inhibited cell viability and growth of EGFR-mutant/osimertinib-resistant cell lines. BLU-945 demonstrated in vivo tumor shrinkage in osimertinib-resistant models of NSCLC (osimertinib second line: EGFR_L858R/C797S and third line: EGFR_ex19del/T790M/C797S and L858R/T790M/C797S) both as monotherapy and in combination with osimertinib. BLU-945 also demonstrated tumor shrinkage in patients from the SYMPHONY trial.
Conclusion: Our findings demonstrate the preclinical and early clinical activity of BLU-945 in EGFRm NSCLC progressing on previous EGFR-TKIs.
{"title":"BLU-945, a potent and selective next-generation EGFR TKI, has antitumor activity in models of osimertinib-resistant non-small-cell lung cancer.","authors":"Sun Min Lim, Stefanie S Schalm, Eun Ji Lee, Sewon Park, Chiara Conti, Yves A Millet, Rich Woessner, Zhuo Zhang, Luz E Tavera-Mendoza, Faith Stevison, Faris Albayya, Thomas A Dineen, John Hsieh, Seung Yeon Oh, Alena Zalutskaya, Julia Rotow, Koichi Goto, Dae-Ho Lee, Mi Ran Yun, Byoung Chul Cho","doi":"10.1177/17588359241280689","DOIUrl":"https://doi.org/10.1177/17588359241280689","url":null,"abstract":"<p><strong>Introduction: </strong>Despite the availability of several epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs), most patients with non-small-cell lung cancer (NSCLC) eventually develop resistance to these agents. Notably, <i>EGFR</i>_C797S mutations confer resistance to the third-generation EGFR-TKI osimertinib and no approved post-osimertinib targeted pharmacology options are currently available. BLU-945 is a novel, reversible, and orally available next-generation EGFR-TKI that selectively targets EGFR-activating (<i>EGFR</i>m) and resistance mutations (including EGFR_C797S) with nanomolar potency while sparing wild-type EGFR in vitro.</p><p><strong>Methods: </strong>In vitro activity of BLU-945 as a single agent and in combination with osimertinib was tested in engineered <i>EGFR</i>-mutant cell lines as well as patient-derived cells and patient-derived organoids. In vivo activity was evaluated in osimertinib-resistant patient-derived xenograft mouse models. Three patient cases from the global, first-in-human, phase I/II SYMPHONY trial (NCT04862780) demonstrating the clinical efficacy of BLU-945 were reported.</p><p><strong>Results: </strong>In vitro BLU-945 demonstrated inhibited cell viability and growth of <i>EGFR</i>-mutant/osimertinib-resistant cell lines. BLU-945 demonstrated in vivo tumor shrinkage in osimertinib-resistant models of NSCLC (osimertinib second line: <i>EGFR</i>_L858R/C797S and third line: <i>EGFR</i>_ex19del/T790M/C797S and L858R/T790M/C797S) both as monotherapy and in combination with osimertinib. BLU-945 also demonstrated tumor shrinkage in patients from the SYMPHONY trial.</p><p><strong>Conclusion: </strong>Our findings demonstrate the preclinical and early clinical activity of BLU-945 in <i>EGFR</i>m NSCLC progressing on previous EGFR-TKIs.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241280689"},"PeriodicalIF":4.3,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11497503/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142508533","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-21eCollection Date: 2024-01-01DOI: 10.1177/17588359241290373
[This corrects the article DOI: 10.1177/17588359241267148.].
[This corrects the article DOI: 10.1177/17588359241267148.].
{"title":"Erratum to \"Additional biomarkers for pathological complete response in triple negative breast cancer\".","authors":"","doi":"10.1177/17588359241290373","DOIUrl":"https://doi.org/10.1177/17588359241290373","url":null,"abstract":"<p><p>[This corrects the article DOI: 10.1177/17588359241267148.].</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241290373"},"PeriodicalIF":4.3,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11497544/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142508535","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-18eCollection Date: 2024-01-01DOI: 10.1177/17588359241289202
Wei Xu, Huai Gong, Bolun Li, Xinmin Yin
Aim: To explore clinical features and prognosis of hepatocellular carcinoma (HCC) in hepatitis B virus surface antigen (HBsAg)-serocleared patients and identify risk factors associated with postoperative recurrence after curative hepatectomy.
Methods: Patients who had undergone initial hepatectomy for HCC from January 2010 through December 2022. Clinicopathological data were compared between HBsAg-seropositive and HBsAg-serocleared patients. Furthermore, risk factors associated with early and late postoperative HCC recurrence (early and late recurrences (ER and LR), respectively) were analyzed for HBsAg-serocleared HCC patients treated by curative hepatectomy.
Results: A total of 2184 consecutive patients undergoing initial hepatectomy for HCC were enrolled, including 339 (15.5%) HBsAg-serocleared and 1845 (84.5%) HBsAg-seropositive cases. Tumor characteristics were comparable between the two groups. After curative hepatectomy, the ER rate was lower in the HBsAg-serocleared group than in the HBsAg-seropositive group (16.2% vs 26.3%; p = 0.000). LR rates in the HBsAg-seropositive and HBsAg-serocleared groups were similar (8.3% vs 6.9%, respectively, p = 0.418). Multivariate analysis showed that among HBsAg-serocleared patients, Hong Kong Liver Cancer stage and microvascular invasion were risk factors associated with postoperative ER, while γ-glutamyl transferase level and neutrophil-to-lymphocyte ratio were associated with LR.
Conclusion: HBsAg-serocleared and HBsAg-seropositive HCC patients exhibited similar tumor characteristics. Curative hepatectomy-treated HBsAg-serocleared HCC patients experienced a lower ER rate and better short-term (⩽3 years) overall survival (OS) rates than their HBsAg-seropositive counterparts. LR, very late recurrence, and long-term (4-, and 5-year) OS rates were similar between the two groups.
{"title":"Hepatocellular carcinoma in HBsAg seroclearance: clinical features, recurrence, and prognosis following curative hepatectomy.","authors":"Wei Xu, Huai Gong, Bolun Li, Xinmin Yin","doi":"10.1177/17588359241289202","DOIUrl":"10.1177/17588359241289202","url":null,"abstract":"<p><strong>Aim: </strong>To explore clinical features and prognosis of hepatocellular carcinoma (HCC) in hepatitis B virus surface antigen (HBsAg)-serocleared patients and identify risk factors associated with postoperative recurrence after curative hepatectomy.</p><p><strong>Methods: </strong>Patients who had undergone initial hepatectomy for HCC from January 2010 through December 2022. Clinicopathological data were compared between HBsAg-seropositive and HBsAg-serocleared patients. Furthermore, risk factors associated with early and late postoperative HCC recurrence (early and late recurrences (ER and LR), respectively) were analyzed for HBsAg-serocleared HCC patients treated by curative hepatectomy.</p><p><strong>Results: </strong>A total of 2184 consecutive patients undergoing initial hepatectomy for HCC were enrolled, including 339 (15.5%) HBsAg-serocleared and 1845 (84.5%) HBsAg-seropositive cases. Tumor characteristics were comparable between the two groups. After curative hepatectomy, the ER rate was lower in the HBsAg-serocleared group than in the HBsAg-seropositive group (16.2% vs 26.3%; <i>p</i> = 0.000). LR rates in the HBsAg-seropositive and HBsAg-serocleared groups were similar (8.3% vs 6.9%, respectively, <i>p</i> = 0.418). Multivariate analysis showed that among HBsAg-serocleared patients, Hong Kong Liver Cancer stage and microvascular invasion were risk factors associated with postoperative ER, while γ-glutamyl transferase level and neutrophil-to-lymphocyte ratio were associated with LR.</p><p><strong>Conclusion: </strong>HBsAg-serocleared and HBsAg-seropositive HCC patients exhibited similar tumor characteristics. Curative hepatectomy-treated HBsAg-serocleared HCC patients experienced a lower ER rate and better short-term (⩽3 years) overall survival (OS) rates than their HBsAg-seropositive counterparts. LR, very late recurrence, and long-term (4-, and 5-year) OS rates were similar between the two groups.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241289202"},"PeriodicalIF":4.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11526261/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142558856","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-18eCollection Date: 2024-01-01DOI: 10.1177/17588359241287658
Loredana G Marcu, David C Marcu
Medical oncology, through conventional chemotherapy as well as targeted drugs, remains an important component of cancer patient management, particularly for systemic disease. Despite advances in all areas of medical oncology, certain challenges persist in the form of drug resistance and severe normal tissue toxicity. These unwanted effects can be counteracted through a patient-tailored treatment approach, which in chemotherapy is translated as pharmacogenomics. This research field investigates the way genetic makeup influences a patient's response to various drugs with the aim to minimize trial-and-error associated with drug administration. The paper introduces the role, advances and challenges of pharmacogenomics, highlighting the importance of Big Data mining to reveal the mechanisms behind drug-gene pair interaction for better patient outcomes. International consortiums have prioritized their focus on the clinical implementation of pharmacogenomics while tackling the challenges ahead: data standardization, ethical aspects and the education of physicians and patients alike to comprehend the power of pharmacogenomics to transform medical oncology.
{"title":"Pharmacogenomics and Big Data in medical oncology: developments and challenges.","authors":"Loredana G Marcu, David C Marcu","doi":"10.1177/17588359241287658","DOIUrl":"10.1177/17588359241287658","url":null,"abstract":"<p><p>Medical oncology, through conventional chemotherapy as well as targeted drugs, remains an important component of cancer patient management, particularly for systemic disease. Despite advances in all areas of medical oncology, certain challenges persist in the form of drug resistance and severe normal tissue toxicity. These unwanted effects can be counteracted through a patient-tailored treatment approach, which in chemotherapy is translated as pharmacogenomics. This research field investigates the way genetic makeup influences a patient's response to various drugs with the aim to minimize trial-and-error associated with drug administration. The paper introduces the role, advances and challenges of pharmacogenomics, highlighting the importance of Big Data mining to reveal the mechanisms behind drug-gene pair interaction for better patient outcomes. International consortiums have prioritized their focus on the clinical implementation of pharmacogenomics while tackling the challenges ahead: data standardization, ethical aspects and the education of physicians and patients alike to comprehend the power of pharmacogenomics to transform medical oncology.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241287658"},"PeriodicalIF":4.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11526290/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142558858","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}